201
|
IFNγ Regulates NAD+ Metabolism to Promote the Respiratory Burst in Human Monocytes. Blood Adv 2022; 6:3821-3834. [PMID: 35500221 DOI: 10.1182/bloodadvances.2021005776] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 04/12/2022] [Indexed: 11/20/2022] Open
Abstract
IFNγ is an essential and pleiotropic activator of human monocytes, but little is known about the changes in cellular metabolism required for IFNγ-induced activation. We sought to elucidate the mechanisms by which IFNγ reprograms monocyte metabolism to support its immunologic activities. We found that IFNγ increased oxygen consumption rates (OCR) in monocytes, indicative of reactive oxygen species generation by both mitochondria and NADPH oxidase. Transcriptional profiling revealed that this oxidative phenotype was driven by IFNγ-induced reprogramming of NAD+ metabolism, which is dependent on nicotinamide phosphoribosyltransferase (NAMPT)-mediated NAD+ salvage to generate NADH and NADPH for oxidation by mitochondrial complex I and NADPH oxidase, respectively. Consistent with this pathway, monocytes from patients with gain-of-function mutations in STAT1 demonstrated higher than normal OCR. Whereas chemical or genetic disruption of mitochondrial complex I (rotenone treatment or Leigh Syndrome patient monocytes) or NADPH oxidase (DPI treatment or chronic granulomatous disease (CGD) patient monocytes) reduced OCR. Interestingly, inhibition of NAMPT in healthy monocytes completely abrogated the IFNγ-induced oxygen consumption, comparable to levels observed in CGD monocytes. These data identify an IFNγ-induced, NAMPT-dependent, NAD+ salvage pathway that is critical for IFNγ activation of human monocytes.
Collapse
|
202
|
Ježek P, Holendová B, Jabůrek M, Dlasková A, Plecitá-Hlavatá L. Contribution of Mitochondria to Insulin Secretion by Various Secretagogues. Antioxid Redox Signal 2022; 36:920-952. [PMID: 34180254 PMCID: PMC9125579 DOI: 10.1089/ars.2021.0113] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Significance: Mitochondria determine glucose-stimulated insulin secretion (GSIS) in pancreatic β-cells by elevating ATP synthesis. As the metabolic and redox hub, mitochondria provide numerous links to the plasma membrane channels, insulin granule vesicles (IGVs), cell redox, NADH, NADPH, and Ca2+ homeostasis, all affecting insulin secretion. Recent Advances: Mitochondrial redox signaling was implicated in several modes of insulin secretion (branched-chain ketoacid [BCKA]-, fatty acid [FA]-stimulated). Mitochondrial Ca2+ influx was found to enhance GSIS, reflecting cytosolic Ca2+ oscillations induced by action potential spikes (intermittent opening of voltage-dependent Ca2+ and K+ channels) or the superimposed Ca2+ release from the endoplasmic reticulum (ER). The ATPase inhibitory factor 1 (IF1) was reported to tune the glucose sensitivity range for GSIS. Mitochondrial protein kinase A was implicated in preventing the IF1-mediated inhibition of the ATP synthase. Critical Issues: It is unknown how the redox signal spreads up to the plasma membrane and what its targets are, what the differences in metabolic, redox, NADH/NADPH, and Ca2+ signaling, and homeostasis are between the first and second GSIS phase, and whether mitochondria can replace ER in the amplification of IGV exocytosis. Future Directions: Metabolomics studies performed to distinguish between the mitochondrial matrix and cytosolic metabolites will elucidate further details. Identifying the targets of cell signaling into mitochondria and of mitochondrial retrograde metabolic and redox signals to the cell will uncover further molecular mechanisms for insulin secretion stimulated by glucose, BCKAs, and FAs, and the amplification of secretion by glucagon-like peptide (GLP-1) and metabotropic receptors. They will identify the distinction between the hub β-cells and their followers in intact and diabetic states. Antioxid. Redox Signal. 36, 920-952.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Blanka Holendová
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin Jabůrek
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Andrea Dlasková
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lydie Plecitá-Hlavatá
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
203
|
Docampo R, Vercesi AE. Mitochondrial Ca 2+ and Reactive Oxygen Species in Trypanosomatids. Antioxid Redox Signal 2022; 36:969-983. [PMID: 34218689 PMCID: PMC9125514 DOI: 10.1089/ars.2021.0058] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/31/2021] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
Significance: Millions of people are infected with trypanosomatids and new therapeutic approaches are needed. Trypanosomatids possess one mitochondrion per cell and its study has led to discoveries of general biological interest. These mitochondria, as in their animal counterparts, generate reactive oxygen species (ROS) and have evolved enzymatic and nonenzymatic defenses against them. Mitochondrial calcium ion (Ca2+) overload leads to generation of ROS and its study could lead to relevant information on the biology of trypanosomatids and to novel drug targets. Recent Advances: Mitochondrial Ca2+ is normally involved in maintaining the bioenergetics of trypanosomes, but when Ca2+ overload occurs, it is associated with cell death. Trypanosomes lack key players in the mechanism of cell death described in mammalian cells, although mitochondrial Ca2+ overload results in collapse of their membrane potential, production of ROS, and cytochrome c release. They are also very resistant to mitochondrial permeability transition, and cell death after mitochondrial Ca2+ overload depends on generation of ROS. Critical Issues: In this review, we consider the mechanisms of mitochondrial oxidant generation and removal and the involvement of Ca2+ in trypanosome cell death. Future Directions: More studies are required to determine the reactions involved in generation of ROS by the mitochondria of trypanosomatids, their enzymatic and nonenzymatic defenses against ROS, and the occurrence and composition of a mitochondrial permeability transition pore. Antioxid. Redox Signal. 36, 969-983.
Collapse
Affiliation(s)
- Roberto Docampo
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | | |
Collapse
|
204
|
Alberici LC, Oliveira HCF. Mitochondrial Adaptive Responses to Hypertriglyceridemia and Bioactive Lipids. Antioxid Redox Signal 2022; 36:953-968. [PMID: 34409856 DOI: 10.1089/ars.2021.0180] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Significance: Altered plasma triglyceride metabolism and changes in dietary fatty acid types and levels are major contributors to the development of metabolic and cardiovascular diseases such as fatty liver disease, obesity, diabetes, and atherosclerosis. Lipid accumulation in visceral adipose tissue and ectopically in other organs, as well as lipid-induced redox imbalance, is connected to mitochondrial dysfunction in a range of oxidative stress-associated metabolic and degenerative disorders. Recent Advances: Successful mitochondrial adaptive responses in the context of hypertriglyceridemia and dietary bioactive polyunsaturated fatty acids contribute to increase body energy expenditure and reduce oxidative stress, thus allowing several cell types to cope with metabolic challenges and stresses. These responses include mitochondrial redox signaling, mild uncoupling, and changes in network dynamic behavior. Critical Issues: Mitochondrial bioenergetics and redox changes in a lipid overload context are relatively well characterized. However, the turning point between adaptive and maladaptive mitochondrial responses remains a critical issue to be elucidated. In addition, the relationship between changes in fusion/fission machinery and mitochondrial function is less well understood. Future Directions: The effective mitochondrial responses described here support the research for new drug design and diet or nutraceutical formulations targeting mitochondrial mild uncoupling and effective quality control as putative strategies for cardiometabolic diseases. Antioxid. Redox Signal. 36, 953-968.
Collapse
Affiliation(s)
- Luciane C Alberici
- Departamento de Ciências BioMoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Helena C F Oliveira
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
205
|
Dawson NJ, Millet C, Selman C, Metcalfe NB. Inter-individual variation in mitochondrial phosphorylation efficiency predicts growth rates in ectotherms at high temperatures. FASEB J 2022; 36:e22333. [PMID: 35486025 DOI: 10.1096/fj.202101806rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 11/11/2022]
Abstract
There is increasing evidence that aquatic ectotherms are especially vulnerable to global warming since their metabolic demands increase with ambient temperature while water-oxygen content decreases. The possible role of shrinking aerobic scope in limiting performance has been much discussed; however, less attention has been given to whether tissue-level changes in the efficiency of oxygen usage occur at elevated temperatures. Here, we show that this varies widely among individuals, with consequences for performance. We examined the inter-individual variation in growth rate and mitochondrial function from white muscle and liver of brown trout (Salmo trutta) acclimated to either high (19.5°C) or near-optimal temperature (12°C). Liver (but not muscle) mitochondria showed a positive relationship between growth rate and maximal oxidative phosphorylation at both temperatures, and a negative relationship between growth rate and ROS release. There was a positive correlation in both tissues between individual mitochondrial phosphorylation efficiency and growth rate, but only at 19.5°C. In this representative of aquatic ectotherms, an individual's liver mitochondrial efficiency thus seems to dictate its capacity to grow at elevated temperatures. This suggests that individual heterogeneity in cellular function may cause variation in the thermal limits of aquatic ectotherms and could adversely affect wild populations in warming environments.
Collapse
Affiliation(s)
- Neal J Dawson
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Caroline Millet
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Colin Selman
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| | - Neil B Metcalfe
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
206
|
Recent Approaches to Determine Static and Dynamic Redox State-Related Parameters. Antioxidants (Basel) 2022; 11:antiox11050864. [PMID: 35624728 PMCID: PMC9137989 DOI: 10.3390/antiox11050864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 11/17/2022] Open
Abstract
Oxidative stress refers to an imbalance between oxidant and antioxidant molecules, which is usually associated with oxidative damage to biomolecules and mitochondrial malfunction. Redox state-related parameters include (1) the direct measurement of ROS, (2) the assessment of the antioxidant defense status, and (3) the analysis of the resulting oxidative damage to molecules. Directly measuring ROS appears to be the preferred method among scientists, but most ROS are extremely unstable and difficult to measure. The processes of determining both the oxidative damage to biomolecules and the antioxidant system status, although both are indirect approaches, provide a reliable method to measure oxidative stress on a given sample. Recently, the Seahorse XF and the Oroboros O2k systems have provided new insights into the redox state from a more dynamic point of view. These techniques assess mitochondrial oxidative phosphorylation function and bioenergetics on isolated mitochondria, cultured cells, or specific tissues such as permeabilized fibers. This review describes a range of methodologies to measure redox state-related parameters, their strengths, and their limitations. In conclusion, all these techniques are valid and none of them can be replaced by another. Indeed, they have the potential to complement each other for a complete evaluation of the redox state of a given sample.
Collapse
|
207
|
Cadenas E. Commentary on "Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria". Arch Biochem Biophys 2022; 726:109214. [PMID: 35483432 DOI: 10.1016/j.abb.2022.109214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
On Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria (Arch Biochem Biophys (1977) 180, 248-257) reviews early work that influenced future studies on the mitochondrial production of superoxide anion and hydrogen peroxide.
Collapse
Affiliation(s)
- Enrique Cadenas
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
208
|
Tomczyk MM, Cheung KG, Xiang B, Tamanna N, Fonseca Teixeira AL, Agarwal P, Kereliuk SM, Spicer V, Lin L, Treberg J, Tong Q, Dolinsky VW. Mitochondrial Sirtuin-3 (SIRT3) Prevents Doxorubicin-Induced Dilated Cardiomyopathy by Modulating Protein Acetylation and Oxidative Stress. Circ Heart Fail 2022; 15:e008547. [PMID: 35418250 PMCID: PMC9117478 DOI: 10.1161/circheartfailure.121.008547] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND High doses of doxorubicin put cancer patients at risk for developing dilated cardiomyopathy. Previously, we showed that doxorubicin treatment decreases SIRT3 (sirtuin 3), the main mitochondrial deacetylase and increases protein acetylation in rat cardiomyocytes. Here, we hypothesize that SIRT3 expression can attenuate doxorubicin induced dilated cardiomyopathy in vivo by preventing the acetylation of mitochondrial proteins. METHODS Nontransgenic, M3-SIRT3 (truncated SIRT3; short isoform), and M1-SIRT3 (full-length SIRT3; mitochondrial localized) transgenic mice were treated with doxorubicin for 4 weeks (8 mg/kg body weight per week). Echocardiography was performed to assess cardiac structure and function and validated by immunohistochemistry and immunofluorescence (n=4-10). Mass spectrometry was performed on cardiac mitochondrial peptides in saline (n=6) and doxorubicin (n=5) treated hearts. Validation was performed in doxorubicin treated primary rat and human induced stem cell derived cardiomyocytes transduced with adenoviruses for M3-SIRT3 and M1-SIRT3 and deacetylase deficient mutants (n=4-10). RESULTS Echocardiography revealed that M3-SIRT3 transgenic mice were partially resistant to doxorubicin induced changes to cardiac structure and function whereas M1-SIRT3 expression prevented cardiac remodeling and dysfunction. In doxorubicin hearts, 37 unique acetylation sites on mitochondrial proteins were altered. Pathway analysis revealed these proteins are involved in energy production, fatty acid metabolism, and oxidative stress resistance. Increased M1-SIRT3 expression in primary rat and human cardiomyocytes attenuated doxorubicin-induced superoxide formation, whereas deacetylase deficient mutants were unable to prevent oxidative stress. CONCLUSIONS Doxorubicin reduced SIRT3 expression and markedly affected the cardiac mitochondrial acetylome. Increased M1-SIRT3 expression in vivo prevented doxorubicin-induced cardiac dysfunction, suggesting that SIRT3 could be a potential therapeutic target for mitigating doxorubicin-induced dilated cardiomyopathy.
Collapse
Affiliation(s)
- Mateusz M Tomczyk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Kyle G Cheung
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Bo Xiang
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Nahid Tamanna
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Ana L Fonseca Teixeira
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Prasoon Agarwal
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,KTH Royal Institute of Technology, School of Electrical Engineering and Computer Science, Stockholm, Sweden (P.A.).,Science for Life Laboratory, Solna, Sweden (P.A.)
| | - Stephanie M Kereliuk
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| | - Victor Spicer
- Department of Internal Medicine (V.S.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada.,Manitoba Center for Proteomics and Systems Biology, Winnipeg, Canada (V.S.)
| | - Ligen Lin
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.).,Institute of Chinese Medical Sciences, University of Macau, China (L.L.)
| | - Jason Treberg
- Department of Biological Sciences (N.T., A.L.F.T., J.T.), University of Manitoba, Winnipeg, Canada
| | - Qiang Tong
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX (L.L., Q.T.)
| | - Vernon W Dolinsky
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme of the Children's Hospital Research Institute of Manitoba' Winnipeg' Canada (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.).,Department of Pharmacology and Therapeutics (M.M.T., K.G.C., B.X., P.A., S.M.K., V.W.D.), University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Science, College of Medicine (M.M.T., K.G.C., B.X., P.A., S.M.K., V.S., V.W.D.), University of Manitoba, Winnipeg, Canada
| |
Collapse
|
209
|
Panov A, Mayorov VI, Dikalov S. Metabolic Syndrome and β-Oxidation of Long-Chain Fatty Acids in the Brain, Heart, and Kidney Mitochondria. Int J Mol Sci 2022; 23:4047. [PMID: 35409406 PMCID: PMC9000033 DOI: 10.3390/ijms23074047] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/04/2023] Open
Abstract
We present evidence that metabolic syndrome (MetS) represents the postreproductive stage of the human postembryonic ontogenesis. Accordingly, the genes governing this stage experience relatively weak evolutionary selection pressure, thus representing the metabolic phenotype of distant ancestors with β-oxidation of long-chain fatty acids (FAs) as the primary energy source. Mitochondria oxidize at high-rate FAs only when succinate, glutamate, or pyruvate are present. The heart and brain mitochondria work at a wide range of functional loads and possess an intrinsic inhibition of complex II to prevent oxidative stress at periods of low functional activity. Kidney mitochondria constantly work at a high rate and lack inhibition of complex II. We suggest that in people with MetS, oxidative stress is the central mechanism of the heart and brain pathologies. Oxidative stress is a secondary pathogenetic mechanism in the kidney, while the primary mechanisms are kidney hypoxia caused by persistent hyperglycemia and hypertension. Current evidence suggests that most of the nongenetic pathologies associated with MetS originate from the inconsistencies between the metabolic phenotype acquired after the transition to the postreproductive stage and excessive consumption of food rich in carbohydrates and a sedentary lifestyle.
Collapse
Affiliation(s)
- Alexander Panov
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31201, USA;
| | - Vladimir I. Mayorov
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, GA 31201, USA;
| | - Sergey Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
210
|
Hoehne MN, Jacobs LJHC, Lapacz KJ, Calabrese G, Murschall LM, Marker T, Kaul H, Trifunovic A, Morgan B, Fricker M, Belousov VV, Riemer J. Spatial and temporal control of mitochondrial H 2 O 2 release in intact human cells. EMBO J 2022; 41:e109169. [PMID: 35146782 PMCID: PMC8982624 DOI: 10.15252/embj.2021109169] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 01/16/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Hydrogen peroxide (H2 O2 ) has key signaling roles at physiological levels, while causing molecular damage at elevated concentrations. H2 O2 production by mitochondria is implicated in regulating processes inside and outside these organelles. However, it remains unclear whether and how mitochondria in intact cells release H2 O2 . Here, we employed a genetically encoded high-affinity H2 O2 sensor, HyPer7, in mammalian tissue culture cells to investigate different modes of mitochondrial H2 O2 release. We found substantial heterogeneity of HyPer7 dynamics between individual cells. We further observed mitochondria-released H2 O2 directly at the surface of the organelle and in the bulk cytosol, but not in the nucleus or at the plasma membrane, pointing to steep gradients emanating from mitochondria. Gradient formation is controlled by cytosolic peroxiredoxins, which act redundantly and with a substantial reserve capacity. Dynamic adaptation of cytosolic thioredoxin reductase levels during metabolic changes results in improved H2 O2 handling and explains previously observed differences between cell types. Our data suggest that H2 O2 -mediated signaling is initiated only in close proximity to mitochondria and under specific metabolic conditions.
Collapse
Affiliation(s)
- Michaela Nicole Hoehne
- Department for ChemistryInstitute for BiochemistryRedox BiochemistryUniversity of CologneCologneGermany
| | - Lianne J H C Jacobs
- Department for ChemistryInstitute for BiochemistryRedox BiochemistryUniversity of CologneCologneGermany
| | - Kim Jasmin Lapacz
- Department for ChemistryInstitute for BiochemistryRedox BiochemistryUniversity of CologneCologneGermany
| | - Gaetano Calabrese
- Department for ChemistryInstitute for BiochemistryRedox BiochemistryUniversity of CologneCologneGermany
| | - Lena Maria Murschall
- Department for ChemistryInstitute for BiochemistryRedox BiochemistryUniversity of CologneCologneGermany
| | - Teresa Marker
- Department for ChemistryInstitute for BiochemistryRedox BiochemistryUniversity of CologneCologneGermany
| | - Harshita Kaul
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Institute for Mitochondrial Diseases and AgingMedical FacultyUniversity of CologneCologneGermany
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
- Institute for Mitochondrial Diseases and AgingMedical FacultyUniversity of CologneCologneGermany
- Center for Molecular MedicineUniversity of CologneCologneGermany
| | - Bruce Morgan
- Institute of BiochemistryCentre for Human and Molecular Biology (ZHMB)Saarland UniversitySaarbrueckenGermany
| | - Mark Fricker
- Department of Plant SciencesUniversity of OxfordOxfordUK
| | - Vsevolod V Belousov
- Department of Metabolism and Redox BiologyShemyakin‐Ovchinnikov Institute of Bioorganic ChemistryMoscowRussia
- Center for Precision Genome Editing and Genetic Technologies for BiomedicinePirogov Russian National Research Medical UniversityMoscowRussia
- Federal Center of Brain Research and NeurotechnologiesFMBAMoscowRussia
- Institute for Cardiovascular PhysiologyGeorg August University GöttingenGöttingenGermany
| | - Jan Riemer
- Department for ChemistryInstitute for BiochemistryRedox BiochemistryUniversity of CologneCologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| |
Collapse
|
211
|
Okoye CN, Chinnappareddy N, Stevens D, Kamunde C. Factors affecting liver mitochondrial hydrogen peroxide emission. Comp Biochem Physiol B Biochem Mol Biol 2022; 259:110713. [PMID: 35026417 DOI: 10.1016/j.cbpb.2022.110713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/19/2021] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
Mitochondria are key cellular sources of reactive oxygen species (ROS) and contain at least 12 known sites on multiple enzymes that convert molecular oxygen to superoxide and hydrogen peroxide (H2O2). Quantitation of site-specific ROS emission is critical to understand the relative contribution of different sites and the pathophysiologic importance of mitochondrial ROS. However, factors that affect mitochondrial ROS emission are not well understood. We characterized and optimized conditions for maximal total and site-specific H2O2 emission during oxidation of standard substrates and probed the source of the high H2O2 emission in unenergized rainbow trout liver mitochondria. We found that mitochondrial H2O2 emission capacity depended on the substrate being oxidized, mitochondrial protein concentration, and composition of the ROS detection system. Contrary to our expectation, addition of exogenous superoxide dismutase reduced H2O2 emission. Titration of conventional mitochondrial electron transfer system (ETS) inhibitors over a range of conditions revealed that one size does not fit all; inhibitor concentrations evoking maximal responses varied with substrate and were moderated by the presence of other inhibitors. Moreover, the efficacy of suppressors of electron leak (S1QEL1.1 and S3QEL2) was low and depended on the substrate being oxidized. We found that H2O2 emission in unenergized rainbow trout liver mitochondria was suppressed by GKT136901 suggesting that it is associated with NADPH oxidase activity. We conclude that optimization of assay conditions is critical for quantitation of maximal H2O2 emission and would facilitate more valid comparisons of mitochondrial total and site-specific H2O2 emission capacities between studies, tissues, and species.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Nirmala Chinnappareddy
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
212
|
Isei MO, Stevens D, Kamunde C. Copper modulates heart mitochondrial H 2O 2 emission differently during fatty acid and pyruvate oxidation. Comp Biochem Physiol C Toxicol Pharmacol 2022; 254:109267. [PMID: 35026399 DOI: 10.1016/j.cbpc.2022.109267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 01/03/2022] [Accepted: 01/05/2022] [Indexed: 12/28/2022]
Abstract
Although the preferred cardiac metabolic fuels are fatty acids, glucose metabolism also plays an important role. However, irrespective of substrate type, energy generation results in mitochondrial reactive oxygen species (ROS) formation. To determine if the preference of fat over carbohydrates predisposes cardiomyocytes to oxidant production, we measured total and site-specific H2O2 emission in heart mitochondria oxidizing palmitoylcarnitine or pyruvate during copper (Cu) exposure. H2O2 emission was higher during oxidation of palmitoylcarnitine compared with pyruvate. Moreover, the bulk of the H2O2 emitted during palmitoylcarnitine oxidation originated from the outer ubiquinone binding site of complex III (site IIIQo) and the flavin site of electron transfer flavoprotein (site EF). We found no evidence of ROS production from complex I ubiquinone-binding site (site IQ) by reverse electron transport during oxidation of palmitoylcarnitine. Pyruvate oxidation also drove H2O2 emission primarily from sites IIIQo; however, the flavin sites of pyruvate dehydrogenase (site PF) and complex II (site IIF) contributed substantially. The effect of Cu depended on substrate and redox site, with effects at sites OF and IIIQo being more pronounced in mitochondria oxidizing pyruvate compared with palmitoylcarnitine. Cu imposed a concentration-saturable effect at site PF but concentration-dependently stimulated H2O2 emission at site EF. The substrate-dependent differences in H2O2 emission and effects of Cu suggest that fuel type and points of entry of electrons into the mitochondrial electron transport system determine the mitochondrial ROS production rate. Importantly, knowledge of sites of mitochondrial ROS production is crucial to the understanding of cardiac dysfunction associated with impaired substrate metabolism.
Collapse
Affiliation(s)
- Michael O Isei
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
213
|
Starr VJ, Dzialowski EM. Developing chicken cardiac muscle mitochondria are resistant to variations in incubation oxygen levels. Curr Res Physiol 2022; 5:151-157. [PMID: 35345510 PMCID: PMC8956876 DOI: 10.1016/j.crphys.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/05/2022] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Chronic exposure to hypoxia during vertebrate development can produce abnormal cardiovascular morphology and function. The aim of this study was to examine cardiac mitochondria function in an avian model, the chicken, in response to embryonic development under hypoxic (15% O2), normoxic (21% O2), or hyperoxic (40% O2) incubation conditions. Methods Chicken embryos were incubated in hypoxia, normoxia, or hyperoxia beginning on day 5 of incubation through hatching. Cardiac mitochondria oxygen flux and reactive oxygen species production were measured in permeabilized cardiac fibers from externally pipped and 1-day post hatchlings. Results Altering oxygen during development had a large effect on body and heart masses of externally pipped embryos and 1-day old hatchlings. Hypoxic animals had smaller body masses and absolute heart masses, but proportionally similar sized hearts compared to normoxic animals during external pipping. Hyperoxic animals were larger with larger hearts than normoxic animals during external pipping. Mitochondrial oxygen flux in permeabilized cardiac muscle fibers revealed limited effects of developing under altered oxygen conditions, with only oxygen flux through cytochrome oxidase being lower in hypoxic hearts compared with hyperoxic hearts. Oxygen flux in leak and oxidative phosphorylation states were not affected by developmental oxygen levels. Mitochondrial reactive oxygen species production under leak and oxidative phosphorylation states studied did not differ between any developmental oxygen treatment. Conclusions These results suggest that cardiac mitochondria function of the developing chicken is not altered by developing in ovo under different oxygen levels. Chicken heart mass is influenced by oxygen availability during development. Cardiac mitochondria respiration was unchanged by developing under hypoxic or hyperoxic oxygen stress. Cardiac mitochondria ROS production was not altered by developmental oxygen stress.
Collapse
Key Words
- AA, Antimycin A
- ADP, adenosine diphosphate
- COX, cytochrome oxidase
- Cardiac mitochondria
- Chicken
- EP, external pipping
- GMP, glutamate, malate, and pyruvate
- Hyperoxia
- Hypoxia
- IP, internal pipping
- LEAK, mitochondrial leak respiration
- OMY, oligomycin
- OXPHOS, mitochondrial oxidative phosphorylation
- ROS, reactive oxygen species
- ROT, rotenone
- Reactive oxygen species
- S, succinate
- TMPD, N,N,N’,N’-tetramethyl-p-phenylenediamine
- dph, days post hatching
Collapse
Affiliation(s)
- Vanessa J Starr
- Developmental Integrative Biology, Department of Biological Sciences, 1155 Union Circle #305220, University of North Texas, Denton, TX, 76203, USA
| | - Edward M Dzialowski
- Developmental Integrative Biology, Department of Biological Sciences, 1155 Union Circle #305220, University of North Texas, Denton, TX, 76203, USA
| |
Collapse
|
214
|
Cadenas E. Commentary on "Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria". Arch Biochem Biophys 2022; 721:109197. [PMID: 35364049 DOI: 10.1016/j.abb.2022.109197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 11/02/2022]
Abstract
On Production of superoxide radicals and hydrogen peroxide by NADH-ubiquinone reductase and ubiquinol-cytochrome c reductase from beef-heart mitochondria (Arch Biochem Biophys (1977) 180, 248-257) reviews early work that influenced future studies on the mitochondrial production of superoxide anion and hydrogen peroxide.
Collapse
Affiliation(s)
- Enrique Cadenas
- Pharmacology & Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
215
|
Feng Y, Luo J, Cheng J, Xu A, Qiu D, He S, Zheng D, Jia C, Zhang Q, Lin N. A Small-Molecule Cocktails-Based Strategy in Culture of Mesenchymal Stem Cells. Front Bioeng Biotechnol 2022; 10:819148. [PMID: 35360405 PMCID: PMC8963903 DOI: 10.3389/fbioe.2022.819148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/11/2022] [Indexed: 12/28/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have a variety of unique properties, such as stem cell multipotency and immune regulation, making them attractive for use in cell therapy. Before infusion therapy, MSCs are required to undergo tissue separation, purification, and expansion in vitro for a certain duration. During the process of in vitro expansion of MSCs, the influence of culture time and environment can lead to cell senescence, increased heterogeneity, and function attenuation, which limits their clinical applications. We used a cocktail of three small-molecule compounds, ACY (A-83–01, CHIR99021, and Y-27632), to increase the proliferation activity of MSCs in vitro and reduce cell senescence. ACY inhibited the increase in heterogeneity of MSCs and conserved their differentiation potential. Additionally, ACY maintained the phenotype of MSCs and upregulated the expression of immunomodulatory factors. These results suggest that ACY can effectively improve the quantity and quality of MSCs.
Collapse
Affiliation(s)
- Yuan Feng
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jintao Cheng
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Aimin Xu
- The First People’s Hospital of Kashi Prefecture, Kashi, China
| | - Dongbo Qiu
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Sixiao He
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dayong Zheng
- The First People’s Hospital of Kashi Prefecture, Kashi, China
| | - Changchang Jia
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| | - Qi Zhang
- Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- *Correspondence: Changchang Jia, ; Qi Zhang, Nan Lin,
| |
Collapse
|
216
|
Rosales M, Rodríguez-Ulloa A, Pérez GV, Besada V, Soto T, Ramos Y, González LJ, Zettl K, Wiśniewski JR, Yang K, Perera Y, Perea SE. CIGB-300-Regulated Proteome Reveals Common and Tailored Response Patterns of AML Cells to CK2 Inhibition. Front Mol Biosci 2022; 9:834814. [PMID: 35359604 PMCID: PMC8962202 DOI: 10.3389/fmolb.2022.834814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/01/2022] [Indexed: 01/13/2023] Open
Abstract
Protein kinase CK2 is a highly pleiotropic and ubiquitously expressed Ser/Thr kinase with instrumental roles in normal and pathological states, including neoplastic phenotype in solid tumor and hematological malignancies. In line with previous reports, CK2 has been suggested as an attractive prognostic marker and molecular target in acute myeloid leukemia (AML), a blood malignant disorder that remains as an unmet medical need. Accordingly, this work investigates the complex landscape of molecular and cellular perturbations supporting the antileukemic effect exerted by CK2 inhibition in AML cells. To identify and functionally characterize the proteomic profile differentially modulated by the CK2 peptide-based inhibitor CIGB-300, we carried out LC-MS/MS and bioinformatic analysis in human cell lines representing two differentiation stages and major AML subtypes. Using this approach, 109 and 129 proteins were identified as significantly modulated in HL-60 and OCI-AML3 cells, respectively. In both proteomic profiles, proteins related to apoptotic cell death, cell cycle progression, and transcriptional/translational processes appeared represented, in agreement with previous results showing the impact of CIGB-300 in AML cell proliferation and viability. Of note, a group of proteins involved in intracellular redox homeostasis was specifically identified in HL-60 cell-regulated proteome, and flow cytometric analysis also confirmed a differential effect of CIGB-300 over reactive oxygen species (ROS) production in AML cells. Thus, oxidative stress might play a relevant role on CIGB-300-induced apoptosis in HL-60 but not in OCI-AML3 cells. Importantly, these findings provide first-hand insights concerning the CIGB-300 antileukemic effect and draw attention to the existence of both common and tailored response patterns triggered by CK2 inhibition in different AML backgrounds, a phenomenon of particular relevance with regard to the pharmacologic blockade of CK2 and personalized medicine.
Collapse
Affiliation(s)
- Mauro Rosales
- Department of Animal and Human Biology, Faculty of Biology, University of Havana (UH), Havana, Cuba
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Arielis Rodríguez-Ulloa
- Mass Spectrometry Laboratory, Proteomics Group, Department of System Biology, Biomedical Research Division, CIGB, Havana, Cuba
| | - George V. Pérez
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Vladimir Besada
- Mass Spectrometry Laboratory, Proteomics Group, Department of System Biology, Biomedical Research Division, CIGB, Havana, Cuba
| | - Thalia Soto
- Department of Animal and Human Biology, Faculty of Biology, University of Havana (UH), Havana, Cuba
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Yassel Ramos
- Mass Spectrometry Laboratory, Proteomics Group, Department of System Biology, Biomedical Research Division, CIGB, Havana, Cuba
| | - Luis J. González
- Mass Spectrometry Laboratory, Proteomics Group, Department of System Biology, Biomedical Research Division, CIGB, Havana, Cuba
| | - Katharina Zettl
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Munich, Germany
| | - Jacek R. Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Munich, Germany
| | - Ke Yang
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd., Yongzhou, China
- *Correspondence: Ke Yang, ; Yasser Perera, ; Silvio E. Perea,
| | - Yasser Perera
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
- China-Cuba Biotechnology Joint Innovation Center (CCBJIC), Yongzhou Zhong Gu Biotechnology Co., Ltd., Yongzhou, China
- *Correspondence: Ke Yang, ; Yasser Perera, ; Silvio E. Perea,
| | - Silvio E. Perea
- Molecular Oncology Group, Department of Pharmaceuticals, Biomedical Research Division, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
- *Correspondence: Ke Yang, ; Yasser Perera, ; Silvio E. Perea,
| |
Collapse
|
217
|
Direct Interaction of Mitochondrial Cytochrome c Oxidase with Thyroid Hormones: Evidence for Two Binding Sites. Cells 2022; 11:cells11050908. [PMID: 35269529 PMCID: PMC8909594 DOI: 10.3390/cells11050908] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 12/18/2022] Open
Abstract
Thyroid hormones regulate tissue metabolism to establish an energy balance in the cell, in particular, by affecting oxidative phosphorylation. Their long-term impact is mainly associated with changes in gene expression, while the short-term effects may differ in their mechanisms. Our work was devoted to studying the short-term effects of hormones T2, T3 and T4 on mitochondrial cytochrome c oxidase (CcO) mediated by direct contact with the enzyme. The data obtained indicate the existence of two separate sites of CcO interaction with thyroid hormones, differing in their location, affinity and specificity to hormone binding. First, we show that T3 and T4 but not T2 inhibit the oxidase activity of CcO in solution and on membrane preparations with Ki ≈ 100–200 μM. In solution, T3 and T4 compete in a 1:1 ratio with the detergent dodecyl-maltoside to bind to the enzyme. The peroxidase and catalase partial activities of CcO are not sensitive to hormones, but electron transfer from heme a to the oxidized binuclear center is affected. We believe that T3 and T4 could be ligands of the bile acid-binding site found in the 3D structure of CcO by Ferguson-Miller’s group, and hormone-induced inhibition is associated with dysfunction of the K-proton channel. A possible role of this interaction in the physiological regulation of the enzyme is discussed. Second, we find that T2, T3, and T4 inhibit superoxide generation by oxidized CcO in the presence of excess H2O2. Inhibition is characterized by Ki values of 0.3–5 μM and apparently affects the formation of O2●− at the protein surface. The second binding site for thyroid hormones presumably coincides with the point of tight T2 binding on the Va subunit described in the literature.
Collapse
|
218
|
Reid MB. Redox Implications of Extreme Task Performance: The Case in Driver Athletes. Cells 2022; 11:cells11050899. [PMID: 35269521 PMCID: PMC8909750 DOI: 10.3390/cells11050899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/26/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
Redox homeostasis and redox-mediated signaling mechanisms are fundamental elements of human biology. Physiological levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) modulate a range of functional processes at the cellular, tissue, and systemic levels in healthy humans. Conversely, excess ROS or RNS activity can disrupt function, impairing the performance of daily activities. This article analyzes the impact of redox mechanisms on extreme task performance. Such activities (a) require complex motor skills, (b) are physically demanding, (c) are performed in an extreme environment, (d) require high-level executive function, and (e) pose an imminent risk of injury or death. The current analysis utilizes race car driving as a representative example. The physiological challenges of this extreme task include physical exertion, g loading, vibration, heat exposure, dehydration, noise, mental demands, and emotional factors. Each of these challenges stimulates ROS signaling, RNS signaling, or both, alters redox homeostasis, and exerts pro-oxidant effects at either the tissue or systemic levels. These redox mechanisms appear to promote physiological stress during race car driving and impair the performance of driver athletes.
Collapse
Affiliation(s)
- Michael B Reid
- College of Health and Human Performance, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
219
|
Koenig A, Buskiewicz-Koenig IA. Redox Activation of Mitochondrial DAMPs and the Metabolic Consequences for Development of Autoimmunity. Antioxid Redox Signal 2022; 36:441-461. [PMID: 35352943 PMCID: PMC8982130 DOI: 10.1089/ars.2021.0073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Reactive oxygen species (ROS) are well known to promote innate immune responses during and in the absence of microbial infections. However, excessive or prolonged exposure to ROS provokes innate immune signaling dysfunction and contributes to the pathogenesis of many autoimmune diseases. The relatively high basal expression of pattern recognition receptors (PRRs) in innate immune cells renders them prone to activation in response to minor intrinsic or extrinsic ROS misbalances in the absence of pathogens. Critical Issues: A prominent source of ROS are mitochondria, which are also major inter-organelle hubs for innate immunity activation, since most PRRs and downstream receptor molecules are directly located either at mitochondria or at mitochondria-associated membranes. Due to their ancestral bacterial origin, mitochondria can also act as quasi-intrinsic self-microbes that mimic a pathogen invasion and become a source of danger-associated molecular patterns (DAMPs) that triggers innate immunity from within. Recent Advances: The release of mitochondrial DAMPs correlates with mitochondrial metabolism changes and increased generation of ROS, which can lead to the oxidative modification of DAMPs. Recent studies suggest that ROS-modified mitochondrial DAMPs possess increased, persistent immunogenicity. Future Directions: Herein, we discuss how mitochondrial DAMP release and oxidation activates PRRs, changes cellular metabolism, and causes innate immune response dysfunction by promoting systemic inflammation, thereby contributing to the onset or progression of autoimmune diseases. The future goal is to understand what the tipping point for DAMPs is to become oxidized, and whether this is a road without return. Antioxid. Redox Signal. 36, 441-461.
Collapse
Affiliation(s)
- Andreas Koenig
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, New York, USA
| | | |
Collapse
|
220
|
da Silva Navarro SM, de Almeida FJS, Luckachaki MD, de Oliveira MR. Sesamol prevents mitochondrial impairment and pro-inflammatory alterations in the human neuroblastoma SH-SY5Y cells: role for Nrf2. Metab Brain Dis 2022; 37:607-617. [PMID: 35000053 DOI: 10.1007/s11011-021-00875-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 11/14/2021] [Indexed: 11/29/2022]
Abstract
Mitochondria are a primary source and a target of reactive oxygen species (ROS). Increased mitochondrial production of ROS is associated with bioenergetics decline, cell death, and inflammation. Here we investigated whether a pretreatment (for 24 h) with sesamol (SES; at 12.5-50 µM) would be efficient in preventing the mitochondrial collapse induced by hydrogen peroxide (H2O2, at 300 µM) in the human neuroblastoma SH-SY5Y cell line. We have found that a pretreatment with SES at 25 µM decreased the effects of H2O2 on lipid peroxidation, protein carbonylation, and protein nitration in membranes obtained from the mitochondria isolated from the SH-SY5Y cells. In this regard, SES pretreatment decreased the production of superoxide anion radical (O2-•) by the mitochondria of H2O2-treated cells. SES also prevented the mitochondrial dysfunction induced by H2O2, as assessed by analyzing the activity of the complexes I and V. The H2O2-induced reduction in the production of adenosine triphosphate (ATP) was also prevented by SES. The levels of the pro-inflammatory cytokines interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α), as well as the activity of the transcription factor nuclear factor-κB (NF-κB) were downregulated by the SES pretreatment in the H2O2-challenged cells. Silencing of the nuclear factor erythroid 2-related factor 2 (Nrf2) transcription factor abolished the protection induced by SES regarding mitochondrial function and inflammation. Thus, SES depends on Nrf2 to promote mitochondrial protection in cells facing redox impairment.
Collapse
Affiliation(s)
- Sônia Mendes da Silva Navarro
- Departamento de Química, Instituto de Ciências Exatas E da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
- Programa de Pós-Graduação Em Química (PPGQ), Universidade Federal de Mato Grosso (UFMT), Cuiabá, MT, Brazil
| | | | - Matheus Dargesso Luckachaki
- Departamento de Química, Instituto de Ciências Exatas E da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil
| | - Marcos Roberto de Oliveira
- Departamento de Química, Instituto de Ciências Exatas E da Terra (ICET), Universidade Federal de Mato Grosso (UFMT), Av. Fernando Corrêa da Costa, 2367, Cuiaba, MT, CEP 78060-900, Brazil.
- Programa de Pós-Graduação Em Química (PPGQ), Universidade Federal de Mato Grosso (UFMT), Cuiabá, MT, Brazil.
| |
Collapse
|
221
|
Zang L, Huang H, Li X, Ju Y, Feng B, Lu J. PEGylated near-infrared fluorescence probe for mitochondria-targetable hydrogen peroxide detection. Talanta 2022; 243:123370. [DOI: 10.1016/j.talanta.2022.123370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/09/2023]
|
222
|
Imdad S, Lim W, Kim JH, Kang C. Intertwined Relationship of Mitochondrial Metabolism, Gut Microbiome and Exercise Potential. Int J Mol Sci 2022; 23:ijms23052679. [PMID: 35269818 PMCID: PMC8910986 DOI: 10.3390/ijms23052679] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
The microbiome has emerged as a key player contributing significantly to the human physiology over the past decades. The potential microbial niche is largely unexplored in the context of exercise enhancing capacity and the related mitochondrial functions. Physical exercise can influence the gut microbiota composition and diversity, whereas a sedentary lifestyle in association with dysbiosis can lead to reduced well-being and diseases. Here, we have elucidated the importance of diverse microbiota, which is associated with an individual's fitness, and moreover, its connection with the organelle, the mitochondria, which is the hub of energy production, signaling, and cellular homeostasis. Microbial by-products, such as short-chain fatty acids, are produced during regular exercise that can enhance the mitochondrial capacity. Therefore, exercise can be employed as a therapeutic intervention to circumvent or subside various metabolic and mitochondria-related diseases. Alternatively, the microbiome-mitochondria axis can be targeted to enhance exercise performance. This review furthers our understanding about the influence of microbiome on the functional capacity of the mitochondria and exercise performance, and the interplay between them.
Collapse
Affiliation(s)
- Saba Imdad
- Molecular Metabolism in Health & Disease, Exercise Physiology Laboratory, Sport Science Research Institute, Inha University, Incheon 22212, Korea;
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Korea
| | - Wonchung Lim
- Department of Sports Medicine, College of Health Science, Cheongju University, Cheongju 28503, Korea;
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, College of Health Science, Cheongju University, Cheongju 28503, Korea
- Correspondence: (J.-H.K.); (C.K.)
| | - Chounghun Kang
- Molecular Metabolism in Health & Disease, Exercise Physiology Laboratory, Sport Science Research Institute, Inha University, Incheon 22212, Korea;
- Department of Physical Education, College of Education, Inha University, Incheon 22212, Korea
- Correspondence: (J.-H.K.); (C.K.)
| |
Collapse
|
223
|
Zhang B, Pan C, Feng C, Yan C, Yu Y, Chen Z, Guo C, Wang X. Role of mitochondrial reactive oxygen species in homeostasis regulation. Redox Rep 2022; 27:45-52. [PMID: 35213291 PMCID: PMC8890532 DOI: 10.1080/13510002.2022.2046423] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are the main source of reactive oxygen species (ROS) in cells. Early studies have shown that mitochondrial reactive oxygen species (mROS) are related to the occurrence and adverse outcomes of many diseases, and are thus regarded as an important risk factor that threaten human health. Recently, increasing evidence has shown that mROS are very important for an organism’s homeostasis. mROS can regulate a variety of signaling pathways and activate the adaptation and protection behaviors of an organism under stress. In addition, mROS also regulate important physiological processes, such as cell proliferation, differentiation, aging, and apoptosis. Herein, we review the mechanisms of production, transformation, and clearance of mROS and their biological roles in different physiological processes.
Collapse
Affiliation(s)
- Baoyi Zhang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
| | - Cunyao Pan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China.,Department of Public Health, Lanzhou University, Lanzhou, People's Republic of China
| | - Chong Feng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China.,School and Hospital of Stomatology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Changqing Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China.,School and Hospital of Stomatology, Tianjin Medical University, Tianjin, People's Republic of China
| | - Yijing Yu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
| | - Zhaoli Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
| | - Changjiang Guo
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
| |
Collapse
|
224
|
Wang X, Ji Y, Jin D, Qi J, Hou X, Zhao W, Zhou S, Zhang C, Luo Y, An P, Luo J. Natural Polysaccharide β-Glucan Protects against Doxorubicin-Induced Cardiotoxicity by Suppressing Oxidative Stress. Nutrients 2022; 14:906. [PMID: 35215555 PMCID: PMC8878312 DOI: 10.3390/nu14040906] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 11/17/2022] Open
Abstract
Doxorubicin (DOXO) can be used to treat a variety of human tumors, but its clinical application is limited due to severe cardiotoxic side effect. Here, we explore the role of β-glucan in DOXO-induced cardiotoxicity in mice and study its underlying mechanism. When co-administered with DOXO, β-glucan was observed to prevent left ventricular dilation and fibrosis. In fact, DOXO reduces the activity of mitochondrial respiratory chain complex and enhances oxidative stress, which in turn impairs heart function. DOXO decreases the ATP production capacity of the heart and increases the ROS content, while β-glucan can restore the heart capacity and reduce oxidative stress. β-glucan also increases the activity of antioxidant enzymes GSH-PX and SOD, and reduces the level of MDA in the serum. In addition, the mRNAs of cardiac dysfunction marker genes ANP, BNP and Myh7 were significantly increased after DOXO induction, however, they did not increase when combined with β-glucan administration. In conclusion, our results indicate that β-glucan can improve the antioxidant capacity of the heart, thereby serving as a potential therapeutic strategy to prevent DOXO-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Yuting Ji
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| | - Dekui Jin
- Department of General Practice, The Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China;
| | - Jingyi Qi
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| | - Xuening Hou
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| | - Wenting Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| | - Shuaishuai Zhou
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| | - Chengying Zhang
- Department of General Practice, The Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China;
| | - Yongting Luo
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| | - Peng An
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| | - Junjie Luo
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.J.); (J.Q.); (X.H.); (W.Z.); (S.Z.)
| |
Collapse
|
225
|
Carpaine Promotes Proliferation and Repair of H9c2 Cardiomyocytes after Oxidative Insults. Pharmaceuticals (Basel) 2022; 15:ph15020230. [PMID: 35215343 PMCID: PMC8880139 DOI: 10.3390/ph15020230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/31/2022] [Accepted: 02/12/2022] [Indexed: 11/22/2022] Open
Abstract
Carpaine has long been identified as the major alkaloid in Carica papaya leaves that possess muscle relaxant properties. Limited study on the molecular signaling properties of carpaine urges us to conduct this study that aims to elucidate the mechanism underlying the cardioprotective effect of carpaine in embryonic cardiomyocytes of the H9c2 cell line. The 50% inhibitory concentration (IC50) of carpaine was first determined using a colorimetric MTT assay to establish the minimum inhibitory concentration for the subsequent test. Using a 1 µM carpaine treatment, a significant increase in the H9c2 proliferation rate was observed following 24 and 48 h of incubation. A Western blot analysis also revealed that carpaine promotes the upregulation of the cell cycle marker proteins cyclin D1 and PCNA. Carpaine-induced H9c2 cell proliferation is mediated by the activation of the FAK-ERK1/2 and FAK-AKT signaling pathways. In the setting of ischemia-reperfusion injury (IRI), carpaine provided a significant protective role to recover the wounded area affected by the hydrogen peroxide (H2O2) treatment. Furthermore, the oxidative-stress-induced reduction in mitochondrial membrane potential (MMP) and overproduction of reactive oxygen species (ROS) were attenuated by carpaine treatment. The current study revealed a novel therapeutic potential of carpaine in promoting in vitro cardiomyocyte proliferation and repair following injury.
Collapse
|
226
|
Jasinski M, Olszewska-Slonina D. Serum Paraoxonase-1 Activity and the Risk of Prostate Cancer Recurrence in Patients Treated with Radiotherapy. Antioxidants (Basel) 2022; 11:antiox11020346. [PMID: 35204228 PMCID: PMC8868428 DOI: 10.3390/antiox11020346] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
The antioxidant paraoxonase-1 (PON1) may be involved in the response to radiation-induced oxidative stress and possibly prevent cell apoptosis. The correlation of PON1 with the risk of cancer recurrence after radiotherapy (RT) is not yet explored. We investigated changes in the activity of PON1 in patients with prostate cancer (PCa) undergoing RT, and the relation of PON1 activity to the risk of recurrence after RT. We included 56 men with PCa. Blood samples were obtained before irradiation and after the completion of RT. Patients were followed for an average of 51.2 months. Each case of biochemical recurrence was confirmed with biopsy. The control group was composed of 60 healthy men. There was no significant difference in PON1 activity between the control group and patients pre-radiotherapy. Irradiation was associated with a significant decrease in PON1 activity. Patients with PCa recurrence had significantly higher serum PON1 activity than those recurrence-free, both before and after RT. PON1 activity was a predictor of PCa recurrence, with sensitivity over 80% and specificity over 64%. Our results suggest that PON1 activity may be a predictor of PCa recurrence risk after RT. Studies with a larger number of patients and longer follow-up are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Milosz Jasinski
- Department of Urology, Institute of Oncology, Romanowskiej 2, 85-796 Bydgoszcz, Poland
- Correspondence:
| | - Dorota Olszewska-Slonina
- Department of Pathobiochemistry and Clinical Chemistry, Collegium Medicum of Nicolaus Copernicus University, M. Curie-Skłodowskiej 9, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
227
|
H2O2/Ca2+/Zn2+ Complex Can Be Considered a “Collaborative Sensor” of the Mitochondrial Capacity? Antioxidants (Basel) 2022; 11:antiox11020342. [PMID: 35204224 PMCID: PMC8868167 DOI: 10.3390/antiox11020342] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
In order to maintain a state of well-being, the cell needs a functional control center that allows it to respond to changes in the internal and surrounding environments and, at the same time, carry out the necessary metabolic functions. In this review, we identify the mitochondrion as such an “agora”, in which three main messengers are able to collaborate and activate adaptive response mechanisms. Such response generators, which we have identified as H2O2, Ca2+, and Zn2+, are capable of “reading” the environment and talking to each other in cooperation with the mitochondrion. In this manner, these messengers exchange information and generate a holistic response of the whole cell, dependent on its functional state. In this review, to corroborate this claim, we analyzed the role these actors, which in the review we call “sensors”, play in the regulation of skeletal muscle contractile capacities chosen as a model of crosstalk between Ca2+, Zn2+, and H2O2.
Collapse
|
228
|
Hirschenson J, Melgar-Bermudez E, Mailloux RJ. The Uncoupling Proteins: A Systematic Review on the Mechanism Used in the Prevention of Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11020322. [PMID: 35204205 PMCID: PMC8868465 DOI: 10.3390/antiox11020322] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/24/2022] [Indexed: 02/04/2023] Open
Abstract
Mitochondrial uncoupling proteins (UCP) 1-3 fulfill many physiological functions, ranging from non-shivering thermogenesis (UCP1) to glucose-stimulated insulin release (GSIS) and satiety signaling (UCP2) and muscle fuel metabolism (UCP3). Several studies have suggested that UCPs mediate these functions by facilitating proton return to the matrix. This would decrease protonic backpressure on the respiratory chain, lowering the production of hydrogen peroxide (H2O2), a second messenger. However, controlling mitochondrial H2O2 production to prevent oxidative stress by activating these leaks through these proteins is still enthusiastically debated. This is due to compelling evidence that UCP2/3 fulfill other function(s) and the inability to reproduce findings that UCP1-3 use inducible leaks to control reactive oxygen species (ROS) production. Further, other studies have found that UCP2/3 may serve as Ca2+. Therefore, we performed a systematic review aiming to summarize the results collected on the topic. A literature search using a list of curated keywords in Pubmed, BIOSIS Citation Index and Scopus was conducted. Potentially relevant references were screened, duplicate references eliminated, and then literature titles and abstracts were evaluated using Rayyan software. A total of 1101 eligible studies were identified for the review. From this total, 416 studies were evaluated based on our inclusion criteria. In general, most studies identified a role for UCPs in preventing oxidative stress, and in some cases, this may be related to the induction of leaks and lowering protonic backpressure on the respiratory chain. However, some studies also generated evidence that UCP2/3 may mitigate oxidative stress by transporting Ca2+ into the matrix, exporting lipid hydroperoxides, or by transporting C-4 metabolites. Additionally, some showed that activating UCP1 or 3 can increase mitochondrial ROS production, even though there is still augmented protection from oxidative stress. Conclusion: Overall, most available studies demonstrate that UCPs, particularly UCP2/3, prevent oxidative stress. However, the mechanism utilized to do so remains elusive and raises the question that UCP2/3 should be renamed, since they may still not be true “uncoupling proteins”.
Collapse
|
229
|
Bekhet OH, Eid ME. The interplay between reactive oxygen species and antioxidants in cancer progression and therapy: a narrative review. Transl Cancer Res 2022; 10:4196-4206. [PMID: 35116715 PMCID: PMC8799102 DOI: 10.21037/tcr-21-629] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022]
Abstract
Objective To unveil the role of reactive oxygen species (ROS) and antioxidants in signaling and involvement in cancer progression and therapy. Background Cancer is considered one of the main causes of mortality in developed countries and expected to be more in developing countries as well. Although some cancers may develop at young age, yet almost all types of cancers are an accumulation of genetic and epigenetic cell damages. Cancer is considered a diverse collection of diseases on a cellular level rather than a single disease; and each disease has a different cause as well. ROS have been seen as harmful toxic molecules; however, they are recognized for cellular signaling capabilities. Elevated levels of ROS have protumorigenic activities; they induce cancer cell proliferation, and adaptation to hypoxia in addition to other effects like DNA damage and genetic instability. They are produced excessively by cancer cells to hyperactivate cellular transformation meanwhile increasing antioxidant capacity to avoid cell death. Methods We discussed peer reviewed published research work from 1987 to 2021. In this paper, we review the role of antioxidants as defensive barrier against excessive ROS levels for maintaining oxidation-reduction (redox) balance; however, antioxidant can also strive in tumor cells with their scavenging capacities and maintain protumorigenic signaling and resist the cancer cell oxidative stress and apoptosis. High doses of antioxidant compounds could be toxic to cells as they are capable of reacting with the physiological concentrations of ROS present for normal cellular processes and signaling. Conclusions Maintaining cellular redox homeostasis is vital for healthy biological system. Therefore, therapeutic modalities for cancer including antioxidants and ROS management should be used at certain doses to target specific redox pathways involved in cancer progression without disrupting the overall redox balance in normal cells.
Collapse
Affiliation(s)
- Osama Hussein Bekhet
- Pole of Endocrinology, Diabetes and Nutrition, Catholic University of Louvain, Woluwe-Saint-Lambert, Belgium
| | - Mohamed Elsayed Eid
- Laboratory of Natural Products Chemistry, Mediterranean Agronomic Institute of Chania, Crete, Greece
| |
Collapse
|
230
|
Is NMDA-Receptor-Mediated Oxidative Stress in Mitochondria of Peripheral Tissues the Essential Factor in the Pathogenesis of Hepatic Encephalopathy? J Clin Med 2022; 11:jcm11030827. [PMID: 35160278 PMCID: PMC8836479 DOI: 10.3390/jcm11030827] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Hepatic encephalopathy (HE) is a neuropsychiatric syndrome of increased ammonia-mediated brain dysfunction caused by impaired hepatic detoxification or when the blood bypasses the liver. Ammonia-activated signal transduction pathways of hyperactivated NMDA receptors (NMDAR) are shown to trigger a cascade of pathological reactions in the brain, leading to oxidative stress. NMDARs outside the brain are widely distributed in peripheral tissues, including the liver, heart, pancreas, and erythrocytes. To determine the contribution of these receptors to ammonia-induced oxidative stress in peripheral tissues, it is relevant to investigate if there are any ammonia-related changes in antioxidant enzymes and free radical formation and whether blockade of NMDARs prevents these changes. Methods: Hyperammonemia was induced in rats by ammonium acetate injection. Oxidative stress was measured as changes in antioxidant enzyme activities and O2•− and H2O2 production by mitochondria isolated from the tissues and cells mentioned above. The effects of the NMDAR antagonist MK-801 on oxidative stress markers and on tissue ammonia levels were evaluated. Results: Increased ammonia levels in erythrocytes and mitochondria isolated from the liver, pancreas, and heart of hyperammonemic rats are shown to cause tissue-specific oxidative stress, which is prevented completely (or partially in erythrocyte) by MK-801. Conclusions: These results support the view that the pathogenesis of HE is multifactorial and that ammonia-induced multiorgan oxidative stress-mediated by activation of NMDAR is an integral part of the disease and, therefore, the toxic effects of ammonia in НЕ may be more global than initially expected.
Collapse
|
231
|
Sun Q, Li Y, Shi L, Hussain R, Mehmood K, Tang Z, Zhang H. Heavy metals induced mitochondrial dysfunction in animals: Molecular mechanism of toxicity. Toxicology 2022; 469:153136. [DOI: 10.1016/j.tox.2022.153136] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 02/11/2022] [Accepted: 02/18/2022] [Indexed: 12/17/2022]
|
232
|
The aryl-ureido fatty acid CTU activates endoplasmic reticulum stress and PERK/NOXA-mediated apoptosis in tumor cells by a dual mitochondrial-targeting mechanism. Cancer Lett 2022; 526:131-141. [PMID: 34822928 DOI: 10.1016/j.canlet.2021.11.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/02/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022]
Abstract
The cancer cell mitochondrion is functionally different from that in normal cells and could be targeted to develop novel experimental therapeutics. The aryl-ureido fatty acid CTU (16({[4-chloro-3-(trifluoromethyl)phenyl]-carbamoyl}amino)hexadecanoic acid) is the prototype of a new class of mitochondrion-targeted agents that kill cancer cells. Here we show that CTU rapidly depolarized the inner mitochondrial membrane, selectively inhibited complex III of the electron transport chain and increased reactive oxygen species (ROS) production. From RNA-seq analysis, endoplasmic reticulum (ER)-stress was a major activated pathway in CTU-treated cells and in MDA-MB-231 tumor xenografts from CTU-treated nu/nu mice. Mitochondrion-derived ROS activated the PERK-linked ER-stress pathway and induced the BH3-only protein NOXA leading to outer mitochondrial membrane (OMM) disruption. The lipid peroxyl scavenger α-tocopherol attenuated CTU-dependent ER-stress and apoptosis which confirmed the critical role of ROS. Oleic acid protected against CTU-mediated apoptosis by activating Mcl-1 expression, which increased NOXA sequestration and prevented OMM disruption. Taken together, CTU both uncouples mitochondrial electron transport and activates ROS production which promotes ER-stress-dependent OMM disruption and tumor cell death. Dual-mitochondrial targeting agents like CTU offer a novel approach for development of new anti-cancer therapeutics.
Collapse
|
233
|
Okoye CN, Chinnappareddy N, Stevens D, Kamunde C. Anoxia-reoxygenation modulates cadmium-induced liver mitochondrial reactive oxygen species emission during oxidation of glycerol 3-phosphate. Comp Biochem Physiol C Toxicol Pharmacol 2022; 252:109227. [PMID: 34728389 DOI: 10.1016/j.cbpc.2021.109227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 11/03/2022]
Abstract
Aquatic organisms are frequently exposed to multiple stressors including low dissolved oxygen (O2) and metals such as cadmium (Cd). Reduced O2 concentration and Cd exposure alter cellular function in part by impairing energy metabolism and dysregulating reactive oxygen species (ROS) homeostasis. However, little is known about the role of mitochondrial glycerol 3-phosphate dehydrogenase (mGPDH) in ROS homeostasis in fish and its response to environmental stress. In this study, mGPDH activity and the effects of anoxia-reoxygenation (A-RO) and Cd on ROS (as hydrogen peroxide, H2O2) emission in rainbow trout liver mitochondria during oxidation of glycerol 3-phosphate (G3P) were probed. Trout liver mitochondria exhibited low mGPDH activity that supported a low respiratory rate but substantial H2O2 emission rate. Cd evoked a low concentration stimulatory-high concentration inhibitory H2O2 emission pattern that was blunted by A-RO. At specific redox centers, Cd suppressed H2O2 emission from site IQ, but stimulated emission from sites IIIQo and GQ. In contrast, A-RO stimulated H2O2 emission from site IQ following 15 min exposure and augmented Cd-stimulated emission from site IIF after 30 min exposure but did not alter the rate of H2O2 emission from sites IIIQo and GQ. Additionally, Cd neither altered the activities of catalase, glutathione peroxidase, or thioredoxin reductase nor the concentrations of total glutathione, reduced glutathione, or oxidized glutathione. Overall, this study indicates that oxidation of G3P drives ROS production from mGPDH and complexes I, II and III, whereas Cd directly modulates redox sites but not antioxidant defense systems to alter mitochondrial H2O2 emission.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Nirmala Chinnappareddy
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Don Stevens
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada
| | - Collins Kamunde
- Department of Biomedical Sciences, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
234
|
Homocysteine-Thiolactone Modulates Gating of Mitochondrial Voltage-Dependent Anion Channel (VDAC) and Protects It from Induced Oxidative Stress. J Membr Biol 2022; 255:79-97. [PMID: 35103807 DOI: 10.1007/s00232-022-00215-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/10/2022] [Indexed: 12/26/2022]
Abstract
The gating of the Voltage-Dependent Anion Channel (VDAC) is linked to oxidative stress through increased generation of mitochondrial ROS with increasing mitochondrial membrane potential (ΔΨm). It has been already reported that H2O2 increases the single-channel conductance of VDAC on a bilayer lipid membrane. On the other hand, homocysteine (Hcy) has been reported to induce mitochondria-mediated cell death. It is argued that the thiol-form of homocysteine, HTL could be the plausible molecule responsible for the alteration in the function of proteins, such as VDAC. It is hypothesized that HTL interacts with VDAC that causes functional abnormalities. An investigation was undertaken to study the interaction of HTL with VDAC under H2O2 induced oxidative stress through biophysical and electrophysiological methods. Fluorescence spectroscopic studies indicate that HTL interacts with VDAC, but under induced oxidative stress the effect is prevented partially. Similarly, bilayer electrophysiology studies suggest that HTL shows a reduction in VDAC single-channel conductance, but the effects are partially prevented under an oxidative environment. Gly172 and His181 are predicted through bioinformatics tools to be the most plausible binding residues of HTL in Rat VDAC. The binding of HTL and H2O2 with VDAC appears to be cooperative as per our analysis of experimental data in the light of the Hill-Langmuir equation. The binding energies are estimated to be - 4.7 kcal mol-1 and - 2.8 kcal mol-1, respectively. The present in vitro studies suggest that when mitochondrial VDAC is under oxidative stress, the effects of amino acid metabolites like HTL are suppressed.
Collapse
|
235
|
Ayer A, Fazakerley DJ, James DE, Stocker R. The role of mitochondrial reactive oxygen species in insulin resistance. Free Radic Biol Med 2022; 179:339-362. [PMID: 34775001 DOI: 10.1016/j.freeradbiomed.2021.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/31/2021] [Accepted: 11/06/2021] [Indexed: 12/21/2022]
Abstract
Insulin resistance is one of the earliest pathological features of a suite of diseases including type 2 diabetes collectively referred to as metabolic syndrome. There is a growing body of evidence from both pre-clinical studies and human cohorts indicating that reactive oxygen species, such as the superoxide radical anion and hydrogen peroxide are key players in the development of insulin resistance. Here we review the evidence linking mitochondrial reactive oxygen species generated within mitochondria with insulin resistance in adipose tissue and skeletal muscle, two major insulin sensitive tissues. We outline the relevant mitochondria-derived reactive species, how the mitochondrial redox state is regulated, and methodologies available to measure mitochondrial reactive oxygen species. Importantly, we highlight key experimental issues to be considered when studying the role of mitochondrial reactive oxygen species in insulin resistance. Evaluating the available literature on both mitochondrial reactive oxygen species/redox state and insulin resistance in a variety of biological systems, we conclude that the weight of evidence suggests a likely role for mitochondrial reactive oxygen species in the etiology of insulin resistance in adipose tissue and skeletal muscle. However, major limitations in the methods used to study reactive oxygen species in insulin resistance as well as the lack of data linking mitochondrial reactive oxygen species and cytosolic insulin signaling pathways are significant obstacles in proving the mechanistic link between these two processes. We provide a framework to guide future studies to provide stronger mechanistic information on the link between mitochondrial reactive oxygen species and insulin resistance as understanding the source, localization, nature, and quantity of mitochondrial reactive oxygen species, their targets and downstream signaling pathways may pave the way for important new therapeutic strategies.
Collapse
Affiliation(s)
- Anita Ayer
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Daniel J Fazakerley
- Metabolic Research Laboratory, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - David E James
- Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Roland Stocker
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia.
| |
Collapse
|
236
|
Qin H, Qu Y, Li R, Qiao J. In Vivo and In Vitro Matured Oocytes From Mice of Advanced Reproductive Age Exhibit Alternative Splicing Processes for Mitochondrial Oxidative Phosphorylation. Front Endocrinol (Lausanne) 2022; 13:816606. [PMID: 35154017 PMCID: PMC8826577 DOI: 10.3389/fendo.2022.816606] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/03/2022] [Indexed: 11/20/2022] Open
Abstract
The mean age of women seeking infertility treatment has gradually increased over recent years. This has coincided with the emergence of in vitro maturation (IVM), a method used in assisted reproductive technology for patients with special requirements. However, when compared with conventional in vitro fertilization, IVM is associated with poor embryonic development potential and low live birth rates, thus limiting the widespread application of this technique. In this study, we performed RNA-sequencing transcriptomic assays and identified a total of 2,627 significant differentially expressed genes (DEGs) between IVM oocytes and in vivo matured oocytes from mice of advanced reproductive age. Next, Kyoto Encyclopedia of Genes and Genomes pathway analysis was used to identify the potential functions of the DEGs. The most significantly enriched pathway was oxidative phosphorylation (OXPHOS). In addition, we constructed a protein-protein interaction network to identify key genes and determined that most of the hub genes were mtDNA-encoded subunits of respiratory chain complex I. Antioxidant supplementation lead to an increase in ATP production and reduced the gene expression profile of the OXPHOS pathway in the IVM group. Moreover, alternative splicing (AS) events were identified during in vivo or in vitro oocyte maturation; data showed that skipped exons were the most frequent type of AS event. A number of genes associated with the OXPHOS pathway exhibited alterations in AS events, including Ndufa7, Ndufs7, Cox6a2, Ndufs5, Ndufb1, and Uqcrh. Furthermore, the process of IVO promoted the skipping of exon 2 in Ndufa7 and exon 3 in Ndufs7 compared with the IVM oocytes, as determined by semi-quantitative RT-PCR. Collectively, these findings provide potential new therapeutic targets for improving IVM of aged women who undergo infertility treatment.
Collapse
Affiliation(s)
- Hao Qin
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yi Qu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
237
|
Xu G, Yuan Y, Luo P, Yang J, Zhou J, Zhu C, Jiang Q, Shu G. Acute Succinate Administration Increases Oxidative Phosphorylation and Skeletal Muscle Explosive Strength via SUCNR1. Front Vet Sci 2022; 8:808863. [PMID: 35097053 PMCID: PMC8795363 DOI: 10.3389/fvets.2021.808863] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/30/2021] [Indexed: 11/15/2022] Open
Abstract
Endurance training and explosive strength training, with different contraction protein and energy metabolism adaptation in skeletal muscle, are both beneficial for physical function and quality of life. Our previous study found that chronic succinate feeding enhanced the endurance exercise of mice by inducing skeletal muscle fiber-type transformation. The purpose of this study is to investigate the effect of acute succinate administration on skeletal muscle explosive strength and its potential mechanism. Succinate was injected to mature mice to explore the acute effect of succinate on skeletal muscle explosive strength. And C2C12 cells were used to verify the short-term effect of succinate on oxidative phosphorylation. Then the cells interfered with succinate receptor 1 (SUCNR1) siRNA, and the SUCNR1-GKO mouse model was used for verifying the role of SUCNR1 in succinate-induced muscle metabolism and expression and explosive strength. The results showed that acute injection of succinate remarkably improved the explosive strength in mice and also decreased the ratio of nicotinamide adenine dinucleotide (NADH) to NAD+ and increased the mitochondrial complex enzyme activity and creatine kinase (CK) activity in skeletal muscle tissue. Similarly, treatment of C2C12 cells with succinate revealed that succinate significantly enhanced oxidative phosphorylation with increased adenosine triphosphate (ATP) content, CK, and the activities of mitochondrial complex I and complex II, but with decreased lactate content, reactive oxygen species (ROS) content, and NADH/NAD+ ratio. Moreover, the succinate's effects on oxidative phosphorylation were blocked in SUCNR1-KD cells and SUCNR1-KO mice. In addition, succinate-induced explosive strength was also abolished by SUCNR1 knockout. All the results indicate that acute succinate administration increases oxidative phosphorylation and skeletal muscle explosive strength in a SUCNR1-dependent manner.
Collapse
|
238
|
Dominiak K, Galganski L, Budzinska A, Woyda-Ploszczyca A, Zoladz JA, Jarmuszkiewicz W. Effects of Endurance Training on the Coenzyme Q Redox State in Rat Heart, Liver, and Brain at the Tissue and Mitochondrial Levels: Implications for Reactive Oxygen Species Formation and Respiratory Chain Remodeling. Int J Mol Sci 2022; 23:ijms23020896. [PMID: 35055078 PMCID: PMC8778810 DOI: 10.3390/ijms23020896] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 02/01/2023] Open
Abstract
Sixteen adult, 4-month-old male Wistar rats were randomly assigned to the training group (n = 8) or the control group (n = 8). We elucidated the effects of 8 weeks of endurance training on coenzyme Q (Q) content and the formation of reactive oxygen species (ROS) at the tissue level and in isolated mitochondria of the rat heart, liver and brain. We demonstrated that endurance training enhanced mitochondrial biogenesis in all tested organs, while a significant increase in the Q redox state was observed in the heart and brain, indicating an elevated level of QH2 as an antioxidant. Moreover, endurance training increased the mQH2 antioxidant pool in the mitochondria of the heart and liver, but not in the brain. At the tissue and isolated mitochondria level, an increase in ROS formation was only observed in the heart. ROS formation observed in the mitochondria of individual rat tissues after training may be associated with changes in the activity/amount of individual components of the oxidative phosphorylation system and its molecular organization, as well as with the size of the oxidized pool of mitochondrial Q acting as an electron carrier in the respiratory chain. Our results indicate that tissue-dependent changes induced by endurance training in the cellular and mitochondrial QH2 pool acting as an antioxidant and in the mitochondrial Q pool serving the respiratory chain may serve important roles in energy metabolism, redox homeostasis and the level of oxidative stress.
Collapse
Affiliation(s)
- Karolina Dominiak
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznan, Poland; (K.D.); (A.B.); (A.W.-P.)
| | - Lukasz Galganski
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznan, Poland; (K.D.); (A.B.); (A.W.-P.)
- Correspondence: (L.G.); (W.J.)
| | - Adrianna Budzinska
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznan, Poland; (K.D.); (A.B.); (A.W.-P.)
| | - Andrzej Woyda-Ploszczyca
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznan, Poland; (K.D.); (A.B.); (A.W.-P.)
| | - Jerzy A. Zoladz
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, 31-066 Krakow, Poland;
| | - Wieslawa Jarmuszkiewicz
- Laboratory of Mitochondrial Biochemistry, Department of Bioenergetics, Faculty of Biology, Adam Mickiewicz University, 61-614 Poznan, Poland; (K.D.); (A.B.); (A.W.-P.)
- Correspondence: (L.G.); (W.J.)
| |
Collapse
|
239
|
Role of Bioactive Compounds in the Regulation of Mitochondrial Dysfunctions in Brain and Age-Related Neurodegenerative Diseases. Cells 2022; 11:cells11020257. [PMID: 35053373 PMCID: PMC8773907 DOI: 10.3390/cells11020257] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondria are multifunctional organelles that participate in a wide range of metabolic processes, including energy production and biomolecule synthesis. The morphology and distribution of intracellular mitochondria change dynamically, reflecting a cell’s metabolic activity. Oxidative stress is defined as a mismatch between the body’s ability to neutralise and eliminate reactive oxygen and nitrogen species (ROS and RNS). A determination of mitochondria failure in increasing oxidative stress, as well as its implications in neurodegenerative illnesses and apoptosis, is a significant developmental process of focus in this review. The neuroprotective effects of bioactive compounds linked to neuronal regulation, as well as related neuronal development abnormalities, will be investigated. In conclusion, the study of secondary components and the use of mitochondrial features in the analysis of various neurodevelopmental diseases has enabled the development of a new class of mitochondrial-targeted pharmaceuticals capable of alleviating neurodegenerative disease states and enabling longevity and healthy ageing for the vast majority of people.
Collapse
|
240
|
A novel sight of the primary active compounds from Umbelliferae: focusing on mitochondria. Med Chem Res 2022. [DOI: 10.1007/s00044-021-02822-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
241
|
Mohd Ghozali N, Giribabu N, Salleh N. Mechanisms Linking Vitamin D Deficiency to Impaired Metabolism: An Overview. Int J Endocrinol 2022; 2022:6453882. [PMID: 35859985 PMCID: PMC9293580 DOI: 10.1155/2022/6453882] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/19/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Vitamin D deficiency is a common health problem worldwide. Despite its known skeletal effects, studies have begun to explore its extra-skeletal effects, that is, in preventing metabolic diseases such as obesity, hyperlipidemia, and diabetes mellitus. The mechanisms by which vitamin D deficiency led to these unfavorable metabolic consequences have been explored. Current evidence indicates that the deficiency of vitamin D could impair the pancreatic β-cell functions, thus compromising its insulin secretion. Besides, vitamin D deficiency could also exacerbate inflammation, oxidative stress, and apoptosis in the pancreas and many organs, which leads to insulin resistance. Together, these will contribute to impairment in glucose homeostasis. This review summarizes the reported metabolic effects of vitamin D, in order to identify its potential use to prevent and overcome metabolic diseases.
Collapse
Affiliation(s)
- Nurulmuna Mohd Ghozali
- Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, Lembah Pantai, Kuala Lumpur 59100, Malaysia
| |
Collapse
|
242
|
Selivanov VA, Zagubnaya OA, Foguet C, Nartsissov YR, Cascante M. MITODYN: An Open Source Software for Quantitative Modeling of Mitochondrial and Cellular Energy Metabolic Flux Dynamics in Health and Disease. Methods Mol Biol 2022; 2399:123-149. [PMID: 35604555 DOI: 10.1007/978-1-0716-1831-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Mitochondrial respiratory chain (RC) transforms the reductive power of NADH or FADH2 oxidation into a proton gradient between the matrix and cytosolic sides of the inner mitochondrial membrane, that ATP synthase uses to generate ATP. This process constitutes a bridge between carbohydrates' central metabolism and ATP-consuming cellular functions. Moreover, the RC is responsible for a large part of reactive oxygen species (ROS) generation that play signaling and oxidizing roles in cells. Mathematical methods and computational analysis are required to understand and predict the possible behavior of this metabolic system. Here we propose a software tool that helps to analyze individual steps of respiratory electron transport in their dynamics, thus deepening understanding of the mechanism of energy transformation and ROS generation in the RC. This software's core is a kinetic model of the RC represented by a system of ordinary differential equations (ODEs). This model enables the analysis of complex dynamic behavior of the RC, including multistationarity and oscillations. The proposed RC modeling method can be applied to study respiration and ROS generation in various organisms and naturally extended to explore carbohydrates' metabolism and linked metabolic processes.
Collapse
Affiliation(s)
- Vitaly A Selivanov
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
- CIBER of Hepatic and Digestive Diseases (CIBEREHD) and Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Institute of Health Carlos III (ISCIII), Madrid, Spain.
| | - Olga A Zagubnaya
- Department of Mathematical Modeling and Statistical Analysis, Institute of Cytochemistry and Molecular Pharmacology, Moscow, Russia
| | - Carles Foguet
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- CIBER of Hepatic and Digestive Diseases (CIBEREHD) and Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Yaroslav R Nartsissov
- Department of Mathematical Modeling and Statistical Analysis, Institute of Cytochemistry and Molecular Pharmacology, Moscow, Russia
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
- CIBER of Hepatic and Digestive Diseases (CIBEREHD) and Metabolomics Node at Spanish National Bioinformatics Institute (INB-ISCIII-ES-ELIXIR), Institute of Health Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
243
|
Kelley RC, Betancourt L, Noriega AM, Brinson SC, Curbelo-Bermudez N, Hahn D, Kumar RA, Balazic E, Muscato DR, Ryan TE, van der Pijl RJ, Shen S, Ottenheijm CAC, Ferreira LF. Skeletal myopathy in a rat model of postmenopausal heart failure with preserved ejection fraction. J Appl Physiol (1985) 2022; 132:106-125. [PMID: 34792407 PMCID: PMC8742741 DOI: 10.1152/japplphysiol.00170.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 11/01/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for ∼50% of all patients with heart failure and frequently affects postmenopausal women. The HFpEF condition is phenotype-specific, with skeletal myopathy that is crucial for disease development and progression. However, most of the current preclinical models of HFpEF have not addressed the postmenopausal phenotype. We sought to advance a rodent model of postmenopausal HFpEF and examine skeletal muscle abnormalities therein. Female, ovariectomized, spontaneously hypertensive rats (SHRs) were fed a high-fat, high-sucrose diet to induce HFpEF. Controls were female sham-operated Wistar-Kyoto rats on a lean diet. In a complementary, longer-term cohort, controls were female sham-operated SHRs on a lean diet to evaluate the effect of strain difference in the model. Our model developed key features of HFpEF that included increased body weight, glucose intolerance, hypertension, cardiac hypertrophy, diastolic dysfunction, exercise intolerance, and elevated plasma cytokines. In limb skeletal muscle, HFpEF decreased specific force by 15%-30% (P < 0.05) and maximal mitochondrial respiration by 40%-55% (P < 0.05), increased oxidized glutathione by approximately twofold (P < 0.05), and tended to increase mitochondrial H2O2 emission (P = 0.10). Muscle fiber cross-sectional area, markers of mitochondrial content, and indices of capillarity were not different between control and HFpEF in our short-term cohort. Overall, our preclinical model of postmenopausal HFpEF recapitulates several key features of the disease. This new model reveals contractile and mitochondrial dysfunction and redox imbalance that are potential contributors to abnormal metabolism, exercise intolerance, and diminished quality of life in patients with postmenopausal HFpEF.NEW & NOTEWORTHY Heart failure with preserved ejection fraction (HFpEF) is a condition with phenotype-specific features highly prevalent in postmenopausal women and skeletal myopathy contributing to disease development and progression. We advanced a rat model of postmenopausal HFpEF with key cardiovascular and systemic features of the disease. Our study shows that the skeletal myopathy of postmenopausal HFpEF includes loss of limb muscle-specific force independent of atrophy, mitochondrial dysfunction, and oxidized shift in redox balance.
Collapse
Affiliation(s)
- Rachel C Kelley
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Lauren Betancourt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Andrea M Noriega
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Suzanne C Brinson
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Nuria Curbelo-Bermudez
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Dongwoo Hahn
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Ravi A Kumar
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Eliza Balazic
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Derek R Muscato
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Terence E Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| | - Robbert J van der Pijl
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
- Department of Physiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Shengyi Shen
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Coen A C Ottenheijm
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
- Department of Physiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| |
Collapse
|
244
|
Duarte-Hospital C, Tête A, Brial F, Benoit L, Koual M, Tomkiewicz C, Kim MJ, Blanc EB, Coumoul X, Bortoli S. Mitochondrial Dysfunction as a Hallmark of Environmental Injury. Cells 2021; 11:cells11010110. [PMID: 35011671 PMCID: PMC8750015 DOI: 10.3390/cells11010110] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 02/07/2023] Open
Abstract
Environmental factors including diet, sedentary lifestyle and exposure to pollutants largely influence human health throughout life. Cellular and molecular events triggered by an exposure to environmental pollutants are extremely variable and depend on the age, the chronicity and the doses of exposure. Only a fraction of all relevant mechanisms involved in the onset and progression of pathologies in response to toxicants has probably been identified. Mitochondria are central hubs of metabolic and cell signaling responsible for a large variety of biochemical processes, including oxidative stress, metabolite production, energy transduction, hormone synthesis, and apoptosis. Growing evidence highlights mitochondrial dysfunction as a major hallmark of environmental insults. Here, we present mitochondria as crucial organelles for healthy metabolic homeostasis and whose dysfunction induces critical adverse effects. Then, we review the multiple mechanisms of action of pollutants causing mitochondrial toxicity in link with chronic diseases. We propose the Aryl hydrocarbon Receptor (AhR) as a model of “exposome receptor”, whose activation by environmental pollutants leads to various toxic events through mitochondrial dysfunction. Finally, we provide some remarks related to mitotoxicity and risk assessment.
Collapse
Affiliation(s)
- Carolina Duarte-Hospital
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Arnaud Tête
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - François Brial
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
| | - Louise Benoit
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Meriem Koual
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Céline Tomkiewicz
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Min Ji Kim
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Université Sorbonne Paris Nord, F-93000 Bobigny, France
| | - Etienne B. Blanc
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
| | - Xavier Coumoul
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
- Correspondence: (X.C.); (S.B.); Tel.: +33-1-76-53-43-70 (S.B.)
| | - Sylvie Bortoli
- Environmental Toxicity, Therapeutic Targets, Cellular Signaling and Biomarkers, T3S, INSERM UMR-S 1124, F-75006 Paris, France; (C.D.-H.); (A.T.); (F.B.); (L.B.); (M.K.); (C.T.); (M.J.K.); (E.B.B.)
- Faculty of Sciences, Université de Paris, F-75006 Paris, France
- Correspondence: (X.C.); (S.B.); Tel.: +33-1-76-53-43-70 (S.B.)
| |
Collapse
|
245
|
Nishida Y, Nawaz A, Hecht K, Tobe K. Astaxanthin as a Novel Mitochondrial Regulator: A New Aspect of Carotenoids, beyond Antioxidants. Nutrients 2021; 14:nu14010107. [PMID: 35010981 PMCID: PMC8746862 DOI: 10.3390/nu14010107] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
Astaxanthin is a member of the carotenoid family that is found abundantly in marine organisms, and has been gaining attention in recent years due to its varied biological/physiological activities. It has been reported that astaxanthin functions both as a pigment, and as an antioxidant with superior free radical quenching capacity. We recently reported that astaxanthin modulated mitochondrial functions by a novel mechanism independent of its antioxidant function. In this paper, we review astaxanthin’s well-known antioxidant activity, and expand on astaxanthin’s lesser-known molecular targets, and its role in mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Yasuhiro Nishida
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Fuji Chemical Industries, Co., Ltd., 55 Yokohoonji, Kamiich-machi, Nakaniikawa-gun, Toyama 930-0405, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| | - Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| | - Karen Hecht
- AstaReal, Inc., 3 Terri Lane, Unit 12, Burlington, NJ 08016, USA;
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| |
Collapse
|
246
|
Flores-Cotera LB, Chávez-Cabrera C, Martínez-Cárdenas A, Sánchez S, García-Flores OU. Deciphering the mechanism by which the yeast Phaffia rhodozyma responds adaptively to environmental, nutritional, and genetic cues. J Ind Microbiol Biotechnol 2021; 48:kuab048. [PMID: 34302341 PMCID: PMC8788774 DOI: 10.1093/jimb/kuab048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/16/2021] [Indexed: 11/13/2022]
Abstract
Phaffia rhodozyma is a basidiomycetous yeast that synthesizes astaxanthin (ASX), which is a powerful and highly valuable antioxidant carotenoid pigment. P. rhodozyma cells accrue ASX and gain an intense red-pink coloration when faced with stressful conditions such as nutrient limitations (e.g., nitrogen or copper), the presence of toxic substances (e.g., antimycin A), or are affected by mutations in the genes that are involved in nitrogen metabolism or respiration. Since cellular accrual of ASX occurs under a wide variety of conditions, this yeast represents a valuable model for studying the growth conditions that entail oxidative stress for yeast cells. Recently, we proposed that ASX synthesis can be largely induced by conditions that lead to reduction-oxidation (redox) imbalances, particularly the state of the NADH/NAD+ couple together with an oxidative environment. In this work, we review the multiple known conditions that elicit ASX synthesis expanding on the data that we formerly examined. When considered alongside the Mitchell's chemiosmotic hypothesis, the study served to rationalize the induction of ASX synthesis and other adaptive cellular processes under a much broader set of conditions. Our aim was to propose an underlying mechanism that explains how a broad range of divergent conditions converge to induce ASX synthesis in P. rhodozyma. The mechanism that links the induction of ASX synthesis with the occurrence of NADH/NAD+ imbalances may help in understanding how other organisms detect any of a broad array of stimuli or gene mutations, and then adaptively respond to activate numerous compensatory cellular processes.
Collapse
Affiliation(s)
- Luis B Flores-Cotera
- Department of Biotechnology and Bioengineering, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, México city 07360, México
| | - Cipriano Chávez-Cabrera
- Department of Biotechnology and Bioengineering, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, México city 07360, México
| | - Anahi Martínez-Cárdenas
- Department of Biotechnology and Bioengineering, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, México city 07360, México
| | - Sergio Sánchez
- Department of Molecular Biology and Biotechnology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México city 04510, México
| | - Oscar Ulises García-Flores
- Department of Biotechnology and Bioengineering, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, México city 07360, México
| |
Collapse
|
247
|
Mookerjee SA, Gerencser AA, Watson MA, Brand MD. Controlled power: how biology manages succinate-driven energy release. Biochem Soc Trans 2021; 49:2929-2939. [PMID: 34882231 PMCID: PMC8786295 DOI: 10.1042/bst20211032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 12/13/2022]
Abstract
Oxidation of succinate by mitochondria can generate a higher protonmotive force (pmf) than can oxidation of NADH-linked substrates. Fundamentally, this is because of differences in redox potentials and gearing. Biology adds kinetic constraints that tune the oxidation of NADH and succinate to ensure that the resulting mitochondrial pmf is suitable for meeting cellular needs without triggering pathology. Tuning within an optimal range is used, for example, to shift ATP consumption between different consumers. Conditions that overcome these constraints and allow succinate oxidation to drive pmf too high can cause pathological generation of reactive oxygen species. We discuss the thermodynamic properties that allow succinate oxidation to drive pmf higher than NADH oxidation, and discuss the evidence for kinetic tuning of ATP production and for pathologies resulting from substantial succinate oxidation in vivo.
Collapse
Affiliation(s)
- Shona A. Mookerjee
- Department of Biological and Pharmaceutical Sciences, Touro University California College of Pharmacy, Vallejo, CA, U.S.A
- Buck Institute for Research on Aging, Novato, CA, U.S.A
| | | | | | - Martin D. Brand
- Department of Biological and Pharmaceutical Sciences, Touro University California College of Pharmacy, Vallejo, CA, U.S.A
- Buck Institute for Research on Aging, Novato, CA, U.S.A
| |
Collapse
|
248
|
Xu Y, Xu Y, Biby S, Bai P, Liu Y, Zhang C, Wang C, Zhang S. Novel Positron Emission Tomography Radiotracers for Imaging Mitochondrial Complex I. ACS Chem Neurosci 2021; 12:4491-4499. [PMID: 34812607 PMCID: PMC10071493 DOI: 10.1021/acschemneuro.1c00297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Mitochondrial dysfunction has been indicated in neurodegenerative and other disorders. The mitochondrial complex I (MC-I) of the electron transport chain (ETC) on the inner membrane is the electron entry point of the ETC and is essential for the production of reactive oxygen species. Based on a recently identified β-keto-amide type MC-I modulator from our laboratory, an 18F-labeled positron emission tomography (PET) tracer, 18F-2, was prepared. PET/CT imaging studies demonstrated that 18F-2 exhibited rapid brain uptake without significant wash out during the 60 min scanning time. In addition, the binding of 18F-2 was higher in the regions of the brain stem, cerebellum, and midbrain. The uptake of 18F-2 can be significantly blocked by its parent compound. Collectively, the results strongly suggest successful development of MC-I PET tracers from this chemical scaffold that can be used in future mitochondrial dysfunction studies of the central nervous system.
Collapse
Affiliation(s)
- Yulong Xu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Yiming Xu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Savannah Biby
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| | - Ping Bai
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Yan Liu
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Can Zhang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Genetics and Aging Research Unit, McCance Center for Brain Health, Mass General Institute for Neurodegenerative Disease, Charlestown, Massachusetts 02129, United States
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia 23298, United States
| |
Collapse
|
249
|
Oxidative distress in aging and age-related diseases: Spatiotemporal dysregulation of protein oxidation and degradation. Biochimie 2021; 195:114-134. [PMID: 34890732 DOI: 10.1016/j.biochi.2021.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/31/2022]
Abstract
The concept of oxidative distress had arisen from the assessment of cellular response to high concentrations of reactive species that result from an imbalance between oxidants and antioxidants and cause biomolecular damage. The intracellular distribution and flux of reactive species dramatically change in time and space contributing to the remodeling of the redox landscape and sensitivity of protein residues to oxidants. Here, we hypothesize that compromised spatiotemporal control of generation, conversions, and removal of reactive species underlies protein damage and dysfunction of protein degradation machineries. This leads to the accumulation of oxidatively damaged proteins resulted in an age-dependent decline in the organismal adaptability to oxidative stress. We highlight recent data obtained with the use of various cell cultures, animal models, and patients on irreversible and non-repairable oxidation of key redox-sensitive residues. Multiple reaction products include peptidyl hydroperoxides, alcohols, carbonyls, and carbamoyl moieties as well as Tyr-Tyr, Trp-Tyr, Trp-Trp, Tyr-Cys, His-Lys, His-Arg, and Tyr-Lys cross-links. These lead to protein fragmentation, misfolding, covalent cross-linking, oligomerization, aggregation, and ultimately, causing impaired protein function and turnover. 20S proteasome and autophagy-lysosome pathways are two major types of machinery for the degradation and elimination of oxidatively damaged proteins. Spatiotemporal dysregulation of these pathways under oxidative distress conditions is implicated in aging and age-related disorders such as neurodegenerative and cardiovascular diseases and diabetes. Future investigations in this field allow the discovery of new drugs to target components of dysregulated cell signaling and protein degradation machinery to combat aging and age-related chronic diseases.
Collapse
|
250
|
López-Aceves TG, Coballase-Urrutia E, Estrada-Rojo F, Vanoye-Carlo A, Carmona-Aparicio L, Hernández ME, Pedraza-Chaverri J, Navarro L, Aparicio-Trejo OE, Pérez-Torres A, Medina-Campos ON, Martínez-Fong D, Sánchez-Valle V, Cárdenas-Rodríguez N, Granados-Rojas L, Pulido-Camarillo E, Rodríguez-Mata V, León-Sicairos CDR. Exposure to Sub-Lethal Doses of Permethrin Is Associated with Neurotoxicity: Changes in Bioenergetics, Redox Markers, Neuroinflammation and Morphology. TOXICS 2021; 9:toxics9120337. [PMID: 34941771 PMCID: PMC8704605 DOI: 10.3390/toxics9120337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 01/15/2023]
Abstract
Permethrin (PERM) is a member of the class I family of synthetic pyrethroids. Human use has shown that it affects different systems, with wide health dysfunctions. Our aim was to determine bioenergetics, neuroinflammation and morphology changes, as redox markers after subacute exposure to PERM in rats. We used MDA determination, protein carbonyl assay, mitochondrial O2 consumption, expression of pro-inflammatory cytokines and a deep histopathological analysis of the hippocampus. PERM (150 mg/kg and 300 mg/kg body weight/day, o.v.) increased lipoperoxidation and carbonylated proteins in a dose-dependent manner in the brain regions. The activities of antioxidant enzymes glutathione peroxidase, reductase, S-transferase, catalase, and superoxide dismutase showed an increase in all the different brain areas, with dose-dependent effects in the cerebellum. Cytokine profiles (IL-1β, IL-6 and TNF-α) increased in a dose-dependent manner in different brain tissues. Exposure to 150 mg/kg of permethrin induced degenerated and/or dead neurons in the rat hippocampus and induced mitochondrial uncoupling and reduction of oxidative phosphorylation and significantly decreased the respiratory parameters state 3-associated respiration in complex I and II. PERM exposure at low doses induces reactive oxygen species production and imbalance in the enzymatic antioxidant system, increases gene expression of pro-inflammatory interleukins, and could lead to cell damage mediated by mitochondrial functional impairment.
Collapse
Affiliation(s)
- Teresita Guadalupe López-Aceves
- Regional Graduate Program in Biotechnology, Faculty of Biological Chemical Sciences, Autonomous University of Sinaloa, Culiacán 80000, Mexico; (T.G.L.-A.); (C.d.R.L.-S.)
- Laboratory of Neuroscience, National Institute of Pediatrics, Mexico City 04530, Mexico; (A.V.-C.); (L.C.-A.); (N.C.-R.); (L.G.-R.)
| | - Elvia Coballase-Urrutia
- Laboratory of Neuroscience, National Institute of Pediatrics, Mexico City 04530, Mexico; (A.V.-C.); (L.C.-A.); (N.C.-R.); (L.G.-R.)
- Correspondence:
| | - Francisco Estrada-Rojo
- Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.E.-R.); (L.N.)
| | - América Vanoye-Carlo
- Laboratory of Neuroscience, National Institute of Pediatrics, Mexico City 04530, Mexico; (A.V.-C.); (L.C.-A.); (N.C.-R.); (L.G.-R.)
| | - Liliana Carmona-Aparicio
- Laboratory of Neuroscience, National Institute of Pediatrics, Mexico City 04530, Mexico; (A.V.-C.); (L.C.-A.); (N.C.-R.); (L.G.-R.)
| | - María Eugenia Hernández
- Subdirection of Clinical Research, National Institute of Psychiatry, Mexico City 14370, Mexico;
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04150, Mexico; (J.P.-C.); (O.E.A.-T.); (O.N.M.-C.)
| | - Luz Navarro
- Department of Physiology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.E.-R.); (L.N.)
| | - Omar E. Aparicio-Trejo
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04150, Mexico; (J.P.-C.); (O.E.A.-T.); (O.N.M.-C.)
| | - Armando Pérez-Torres
- Department of Cell and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.P.-T.); (E.P.-C.); (V.R.-M.)
| | - Omar N. Medina-Campos
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04150, Mexico; (J.P.-C.); (O.E.A.-T.); (O.N.M.-C.)
| | - Daniel Martínez-Fong
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies, Mexico City 07360, Mexico;
| | - Vicente Sánchez-Valle
- Neuroplasticity and Neurodegeneration Laboratory, Department of Pharmacology, Center for Research and Advanced Studies, Mexico City 07360, Mexico;
| | - Noemi Cárdenas-Rodríguez
- Laboratory of Neuroscience, National Institute of Pediatrics, Mexico City 04530, Mexico; (A.V.-C.); (L.C.-A.); (N.C.-R.); (L.G.-R.)
| | - Leticia Granados-Rojas
- Laboratory of Neuroscience, National Institute of Pediatrics, Mexico City 04530, Mexico; (A.V.-C.); (L.C.-A.); (N.C.-R.); (L.G.-R.)
| | - Evelyn Pulido-Camarillo
- Department of Cell and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.P.-T.); (E.P.-C.); (V.R.-M.)
| | - Verónica Rodríguez-Mata
- Department of Cell and Tissue Biology, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.P.-T.); (E.P.-C.); (V.R.-M.)
| | - Claudia del R. León-Sicairos
- Regional Graduate Program in Biotechnology, Faculty of Biological Chemical Sciences, Autonomous University of Sinaloa, Culiacán 80000, Mexico; (T.G.L.-A.); (C.d.R.L.-S.)
| |
Collapse
|